1
|
Guan Q, Gilpin SG, Doerksen J, Bath L, Lam T, Li Y, Lambert P, Wall DA. The Interactions of T Cells with Myeloid-Derived Suppressor Cells in Peripheral Blood Stem Cell Grafts. Cells 2024; 13:1545. [PMID: 39329729 PMCID: PMC11429538 DOI: 10.3390/cells13181545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
The interaction of myeloid-derived suppressor cells (MDSCs) with T cells within G-CSF-mobilized peripheral blood stem cell (PBSC) grafts in patients undergoing autologous or allogeneic hematopoietic stem cell transplantation remains to be elucidated. Through studying allo- and auto-PBSC grafts, we observed grafts containing large numbers of T cells and MDSCs with intergraft variability in their percentage and number. T cells from autologous grafts compared to allografts expressed relative higher percentages of inhibitory receptors PD-1, CTLA-4, TIM-3, LAG-3, TIGIT and BTLA. Autograft T cells had decreased cell proliferation and IFN-γ secretion, which supported the possible presence of T cell exhaustion. On the contrary, graft monocytic MDSCs (M-MDSCs) expressed multiple inhibitory receptor ligands, including PD-L1, CD86, Galectin-9, HVEM and CD155. The expression of inhibitory receptor ligands on M-MDSCs was correlated with their corresponding inhibitory receptors on T cells in the grafts. Isolated M-MDSCs had the ability to suppress T cell proliferation and IFN-γ secretion and/or promote Treg expansion. Blocking the PD-L1-PD-1 signaling pathway partially reversed the functions of M-MDSCs. Taken together, our data indicated that T cells and M-MDSCs in PBSC grafts express complementary inhibitory receptor-ligand pairing, which may impact the quality of immune recovery and clinical outcome post transplantation.
Collapse
Affiliation(s)
- Qingdong Guan
- Manitoba Blood and Marrow Transplant Program, Departments of Pediatrics and Child Health and Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada (D.A.W.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Manitoba Center for Advanced Cell and Tissue Therapy, Winnipeg, MB R3A 1R9, Canada
- Paul Albreachtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3A 1R9, Canada
| | - Scott G. Gilpin
- Manitoba Blood and Marrow Transplant Program, Departments of Pediatrics and Child Health and Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada (D.A.W.)
| | - James Doerksen
- Manitoba Blood and Marrow Transplant Program, Departments of Pediatrics and Child Health and Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada (D.A.W.)
| | - Lauren Bath
- Manitoba Blood and Marrow Transplant Program, Departments of Pediatrics and Child Health and Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada (D.A.W.)
| | - Tracy Lam
- Manitoba Blood and Marrow Transplant Program, Departments of Pediatrics and Child Health and Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada (D.A.W.)
| | - Yun Li
- Manitoba Blood and Marrow Transplant Program, Departments of Pediatrics and Child Health and Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada (D.A.W.)
| | - Pascal Lambert
- Department of Epidemiology and Cancer Registry, CancerCare Manitoba, Winnipeg, MB R3A 1R9, Canada;
| | - Donna A. Wall
- Manitoba Blood and Marrow Transplant Program, Departments of Pediatrics and Child Health and Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada (D.A.W.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Manitoba Center for Advanced Cell and Tissue Therapy, Winnipeg, MB R3A 1R9, Canada
- Paul Albreachtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3A 1R9, Canada
| |
Collapse
|
2
|
Al-Ibraheem A, Mottaghy FM, Juweid ME. PET/CT in Hodgkin Lymphoma: An Update. Semin Nucl Med 2023; 53:303-319. [PMID: 36369090 DOI: 10.1053/j.semnuclmed.2022.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022]
Abstract
18F-FDG-PET/CT is now an integral part of the workup and management of patients with Hodgkin's lymphoma (HL). PET/CT is currently routinely performed for staging and for response assessment at the end of treatment. Interim PET/CT is typically performed after 1-4 of 6-8 chemo/chemoimmunotherapy cycles ± radiation for prognostication and potential treatment escalation or de-escalation early in the course of therapy, a concept known as response-or risk-adapted treatment. Quantitative PET is an area of growing interest. Metrics such as the standardized uptake value (SUV), metabolic tumor volume, total lesion glycolysis, and their changes with treatment are being investigated as more reproducible and, potentially, more accurate predictors of response and prognosis. Despite the progress made in standardizing the use of PET/CT in lymphoma, challenges remain, particularly with respect to its limited positive predictive value. This review highlights the most relevant applications of PET/CT in HL, its strengths and limitations, as well as recent efforts to implement PET/CT-based metrics as promising tools for precision medicine. Finally, the value of PET/CT for response assessment to immunotherapy is discussed.
Collapse
Affiliation(s)
- Akram Al-Ibraheem
- Department of Nuclear Medicine, King Hussein Cancer Center, Amman, Jordan; Division of Nuclear Medicine/Department of Radiology and Nuclear Medicine, University of Jordan, Amman, Jordan
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH, Aachen University, Aachen, 52074, Germany, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany and Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.
| | - Malik E Juweid
- Division of Nuclear Medicine/Department of Radiology and Nuclear Medicine, University of Jordan, Amman, Jordan
| |
Collapse
|
3
|
Ke H, Zhang F, Wang J, Xiong L, An X, Tu X, Chen C, Wang Y, Mao B, Guo S, Ju C, He X, Sun R, Zhang L, O'Connor OA, Li QX. HX009, a novel BsAb dual targeting PD1 x CD47, demonstrates potent anti-lymphoma activity in preclinical models. Sci Rep 2023; 13:5419. [PMID: 37012357 PMCID: PMC10070465 DOI: 10.1038/s41598-023-32547-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Both PD1/PD-L1 and CD47 blockades have demonstrated limited activity in most subtypes of NHL save NK/T-cell lymphoma. The hemotoxicity with anti-CD47 agents in the clinic has been speculated to account for their limitations. Herein we describe a first-in-class and rationally designed bispecific antibody (BsAb), HX009, targeting PD1 and CD47 but with weakened CD47 binding, which selectively hones the BsAb for tumor microenvironment through PD1 interaction, potentially reducing toxicity. In vitro characterization confirmed: (1) Both receptor binding/ligand blockade, with lowered CD47 affinity; (2) functional PD1/CD47 blockades by reporter assays; (3) T-cell activation in Staphylococcal-enterotoxin-B-pretreated PBMC and mixed-lymphocyte-reaction. In vivo modeling demonstrated antitumor activity in Raji-B and Karpass-229-T xenograft lymphomas. In the humanized mouse syngeneic A20 B-lymphoma (huCD47-A20) HuGEMM model, which has quadruple knocked-in hPD1xhPD-L1xhCD47xhSIRPα genes and an intact autologous immune-system, a contribution of effect is demonstrated for each targeted biologic (HX008 targeting PD1 and SIRPα-Fc targeting CD47), which is clearly augmented by the dual targeting with HX009. Lastly, the expression of the immune-checkpoints PD-L1/L2 and CD47 seemed co-regulated among a panel of lymphoma-derived-xenografts, where HX009 maybe more effective in those with upregulated CD47. Our data warrants HX009's further clinical development for treating NHLs.
Collapse
Affiliation(s)
- Hang Ke
- Hanx Pharmaceuticals, Inc., Hangzhou, China
| | | | | | | | - Xiaoyu An
- Crown Bioscience, Inc., San Diego, USA
| | | | - Cen Chen
- Hanx Pharmaceuticals, Inc., Hangzhou, China
| | | | | | - Sheng Guo
- Crown Bioscience, Inc., San Diego, USA
| | | | - Xiangfei He
- Shanghai Model Organisms Center, Inc. (SMOC), Shanghai, China
| | - Ruilin Sun
- Shanghai Model Organisms Center, Inc. (SMOC), Shanghai, China
| | - Lei Zhang
- Hanx Pharmaceuticals, Inc., Hangzhou, China
| | - Owen A O'Connor
- Division of Hematology and Oncology, University of Virginia Cancer Center, University of Virginia, Charlottesville, USA
| | - Qi-Xiang Li
- Hanx Pharmaceuticals, Inc., Hangzhou, China.
| |
Collapse
|
4
|
Henry M, Buck S, Al-Qanber B, Gadgeel M, Savaşan S. Lymphocyte HLA-DR/CD-38 co-expression correlates with Hodgkin lymphoma cell cytotoxicity in vitro independent of PD-1/PD1-L pathway. Leuk Lymphoma 2022; 63:1331-1338. [PMID: 35001800 DOI: 10.1080/10428194.2021.2023744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The interactions between Hodgkin and Reed Sternberg cells and tumor microenvironment, the changes that occur with therapy and, in particular, checkpoint inhibition are not fully understood. Understanding these is key to optimizing outcomes for patients with Hodgkin lymphoma (HL). We evaluated the immunophenotypic characteristics of cytotoxic, helper T and NK lymphocytes upon in vitro stimulation, cell-mediated cytotoxicity against HL cells, HDLM-2 and KM-H2, and the association with effector cell activation state, as well as changes in cytotoxicity following PD-1 or PDL-1 blockade. Higher HLA-DR/CD38 expression on effector cells was associated with increased cytotoxicity against HL cells. All effector cell types were cytotoxic of HL cells, though achieved maximum activation and cytotoxicity at variable timepoints. HLA-DR/CD38 co-expression correlated with cytotoxicity, but PD-1 expression did not. There was no significant change in cell-mediated cytotoxicity following PD-1/PDL-1 blockade. The mechanism of action of checkpoint inhibitors may not be limited to direct PD-1/PDL-1 blockade.
Collapse
Affiliation(s)
- Meret Henry
- Division of Hematology/Oncology, Pediatric Blood and Marrow Transplant Program, Barbara Ann Karmanos Cancer Center, Children's Hospital of Michigan, Detroit, MI, USA.,Department of Pediatrics, Central Michigan University College of Medicine, Mount Pleasant, MI, USA
| | - Steven Buck
- Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Batool Al-Qanber
- Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Manisha Gadgeel
- Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Süreyya Savaşan
- Division of Hematology/Oncology, Pediatric Blood and Marrow Transplant Program, Barbara Ann Karmanos Cancer Center, Children's Hospital of Michigan, Detroit, MI, USA.,Department of Pediatrics, Central Michigan University College of Medicine, Mount Pleasant, MI, USA.,Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| |
Collapse
|
5
|
Essential role of the linear ubiquitin chain assembly complex and TAK1 kinase in A20 mutant Hodgkin lymphoma. Proc Natl Acad Sci U S A 2020; 117:28980-28991. [PMID: 33139544 DOI: 10.1073/pnas.2014470117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
More than 70% of Epstein-Barr virus (EBV)-negative Hodgkin lymphoma (HL) cases display inactivation of TNFAIP3 (A20), a ubiquitin-editing protein that regulates nonproteolytic protein ubiquitination, indicating the significance of protein ubiquitination in HL pathogenesis. However, the precise mechanistic roles of A20 and the ubiquitination system remain largely unknown in this disease. Here, we performed high-throughput CRISPR screening using a ubiquitin regulator-focused single-guide RNA library in HL lines carrying either wild-type or mutant A20. Our CRISPR screening highlights the essential oncogenic role of the linear ubiquitin chain assembly complex (LUBAC) in HL lines, which overlaps with A20 inactivation status. Mechanistically, LUBAC promotes IKK/NF-κB activity and NEMO linear ubiquitination in A20 mutant HL cells, which is required for prosurvival genes and immunosuppressive molecule expression. As a tumor suppressor, A20 directly inhibits IKK activation and HL cell survival via its C-terminal linear-ubiquitin binding ZF7. Clinically, LUBAC activity is consistently elevated in most primary HL cases, and this is correlated with high NF-κB activity and low A20 expression. To further understand the complete mechanism of NF-κB activation in A20 mutant HL, we performed a specifically designed CD83-based NF-κB CRISPR screen which led us to identify TAK1 kinase as a major mediator for NF-κB activation in cells dependent on LUBAC, where the LUBAC-A20 axis regulates TAK1 and IKK complex formation. Finally, TAK1 inhibitor Takinib shows promising activity against HL in vitro and in a xenograft mouse model. Altogether, these findings provide strong support that targeting LUBAC or TAK1 could be attractive therapeutic strategies in A20 mutant HL.
Collapse
|
6
|
Diefenbach CS, Hong F, Ambinder RF, Cohen JB, Robertson MJ, David KA, Advani RH, Fenske TS, Barta SK, Palmisiano ND, Svoboda J, Morgan DS, Karmali R, Sharon E, Streicher H, Kahl BS, Ansell SM. Ipilimumab, nivolumab, and brentuximab vedotin combination therapies in patients with relapsed or refractory Hodgkin lymphoma: phase 1 results of an open-label, multicentre, phase 1/2 trial. LANCET HAEMATOLOGY 2020; 7:e660-e670. [PMID: 32853585 DOI: 10.1016/s2352-3026(20)30221-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recognising that the immune suppressive microenvironment promotes tumour growth in Hodgkin lymphoma, we hypothesised that activating immunity might augment the activity of targeted chemotherapy. We evaluated the safety and activity of combinations of brentuximab vedotin with nivolumab or ipilimumab, or both in patients with relapsed or refractory Hodgkin lymphoma. METHODS In this multicentre, open-label, phase 1/2 trial, patients with relapsed or refractory Hodgkin lymphoma aged 18 years or older who had relapsed after at least one line of therapy, with an Eastern Cooperative Oncology Group performance status of 2 or lower, and adequate organ and marrow function, with no pulmonary dysfunction were eligible for inclusion. Phase 1 primary objectives were to determine the maximum tolerated dose and dose limiting toxicities of brentuximab vedotin combined with ipilimumab (ipilimumab group), nivolumab (nivolumab group), or both (triplet therapy group) using a 3 + 3 dose escalation design with expansion cohorts. During the dose escalation phase, patients were enrolled sequentially into one of six cohorts: in the ipilimumab group fixed brentuximab vedotin 1·8 mg/kg with ipilimumab 1 mg/kg (cohort A) or 3 mg/kg (cohort B); in the nivolumab group fixed nivolumab 3 mg/kg with brentuximab vedotin 1·2 mg/kg (cohort D) or 1·8 mg/kg (cohort E); and in the triplet therapy group fixed nivolumab 3 mg/kg and ipilimumab 1 mg/kg with brentuximab vedotin 1·2 mg/kg (cohort G) or 1·8 mg/kg (cohort H). Additional patients were enrolled in the expansion phase at the same doses of cohorts B, E, and H. All drugs were given intravenously; brentuximab vedotin and nivolumab were given every 3 weeks, ipilimumab was given every 6 weeks in the ipilimumab group and every 12 weeks in the triplet therapy group. All eligible and treated patients were included in the analysis. This phase 1/2 study is registered with ClinicalTrials.gov, NCT01896999. The phase 2, randomised portion of the trial is still enrolling. FINDINGS Between March 7, 2014, and Dec 28, 2017, 64 patients were enrolled; two patients in the ipilimumab group and one patient in the nivolumab group were excluded due to ineligibility after enrolment and 61 were evaluable. A total of six dose limiting toxicities were reported in four patients, and the doses used in cohorts B, E, and H were established as maximum tolerated doses and patients were subsequently enrolled onto expansion cohorts (C, F, and I) with these schedules. There were ten (43%) grade 3-4 treatment related adverse events in the ipilimumab group, three (16%) in the nivolumab group, and 11 (50%) in the triplet therapy group including: eight (13%) of 64 patients reporting rash, and colitis, gastritis, pancreatitis and arthritis, and diabetic ketoacidosis each occurring in one (2%) patient. There were two (3%) treatment related deaths, one in the nivolumab group and one in the triplet therapy group. The overall response rate was 76% (95% CI 53-92) in the ipilimumab group, 89% (65-99) in the nivolumab group, and 82% (60-95) in the triplet therapy group, and the complete response rate was 57% (95% CI 34-78%) in the ipilimumab group, 61% (36-83%) in the nivolumab group, and 73% (50-89%) in the triplet therapy group. With a median follow-up of 2·6 years (IQR 1·8-2·9) in the ipilimumab group, 2·4 years (2·2-2·6) in the nivolumab group, and 1·7 years (1·6-1·9) in the triplet therapy group, median progression-free survival is 1·2 years (95% CI 1·7-not reached) in the ipilimumab group, but was not reached in the other two treatment groups. Median overall survival has not been reached in any of the groups. INTERPRETATION There are clear differences in activity and toxicity of the three combination regimens. The tolerability and preliminary activity for the two most active regimens, brentuximab vedotin with nivolumab and the triplet therapy, are being compared in a randomised phase 2 trial (NCT01896999). FUNDING Eastern Cooperative Oncology Group-American College of Radiology Imaging Network and the National Cancer Institute of the National Institutes of Health.
Collapse
Affiliation(s)
| | - Fangxin Hong
- Eastern Cooperative Oncology Group-American College of Radiology Imaging Network Biostatistics Center, Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Kevin A David
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Jersey, NJ, USA
| | | | - Timothy S Fenske
- Division of Hematology and Oncology, Froedtert and the Medical College of Wisconsin, Wauwatosa, WI, USA
| | - Stefan K Barta
- Divison of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil D Palmisiano
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jakub Svoboda
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - David S Morgan
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Reem Karmali
- Robert H Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, USA
| | - Elad Sharon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethseda, MD, USA
| | - Howard Streicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethseda, MD, USA
| | - Brad S Kahl
- School of Medicine, Washington University, St Louis, MO, USA
| | | |
Collapse
|
7
|
Lopci E, Meignan M. Current Evidence on PET Response Assessment to Immunotherapy in Lymphomas. PET Clin 2020; 15:23-34. [DOI: 10.1016/j.cpet.2019.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
8
|
Mendez LM, Posey RR, Pandolfi PP. The Interplay Between the Genetic and Immune Landscapes of AML: Mechanisms and Implications for Risk Stratification and Therapy. Front Oncol 2019; 9:1162. [PMID: 31781488 PMCID: PMC6856667 DOI: 10.3389/fonc.2019.01162] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022] Open
Abstract
AML holds a unique place in the history of immunotherapy by virtue of being among the first malignancies in which durable remissions were achieved with "adoptive immunotherapy," now known as allogeneic stem cell transplantation. The successful deployment of unselected adoptive cell therapy established AML as a disease responsive to immunomodulation. Classification systems for AML have been refined and expanded over the years in an effort to capture the variability of this heterogeneous disease and risk-stratify patients. Current systems increasingly incorporate information about cytogenetic alterations and genetic mutations. The advent of next generation sequencing technology has enabled the comprehensive identification of recurrent genetic mutations, many with predictive power. Recurrent genetic mutations found in AML have been intensely studied from a cell intrinsic perspective leading to the genesis of multiple, recently approved targeted therapies including IDH1/2-mutant inhibitors and FLT3-ITD/-TKD inhibitors. However, there is a paucity of data on the effects of these targeted agents on the leukemia microenvironment, including the immune system. Recently, the phenomenal success of checkpoint inhibitors and CAR-T cells has re-ignited interest in understanding the mechanisms leading to immune dysregulation and suppression in leukemia, with the objective of harnessing the power of the immune system via novel immunotherapeutics. A paradigm has emerged that places crosstalk with the immune system at the crux of any effective therapy. Ongoing research will reveal how AML genetics inform the composition of the immune microenvironment paving the way for personalized immunotherapy.
Collapse
Affiliation(s)
- Lourdes M. Mendez
- Department of Medicine and Pathology, Cancer Research Institute, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, United States
| | - Ryan R. Posey
- Department of Medicine and Pathology, Cancer Research Institute, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, United States
| | - Pier Paolo Pandolfi
- Department of Medicine and Pathology, Cancer Research Institute, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
9
|
Abstract
Classic Hodgkin lymphoma has a unique tumor composition in that there is a paucity of malignant cells present, and most of the tumor consists of normal immune and stromal cells. Despite the presence of an immune infiltrate within the tumor microenvironment, the malignant cells effectively evade the immune system and appear to utilize the presence of immune cells to promote the growth and survival of Hodgkin-Reed-Sternberg cells. Hodgkin-Reed-Sternberg cells evade immune detection because of overexpression of programmed death 1 ligands, PD-L1 and PD-L2, which suppress T-cell activation, and loss of expression of major histocompatibility complex molecules that prevent effective immune recognition. Recognition of these immune defects has led to clinical use of immune checkpoint blockade in classic Hodgkin lymphoma. Clinical trials using antibodies that block programmed death 1/PD-L1 signaling have shown remarkable responses to therapy and have led to the approval of nivolumab and pembrolizumab for use in patients with relapsed and refractory disease. Trials are currently testing immune checkpoint blockade in earlier lines of therapy.
Collapse
|
10
|
Aldinucci D, Borghese C, Casagrande N. Formation of the Immunosuppressive Microenvironment of Classic Hodgkin Lymphoma and Therapeutic Approaches to Counter It. Int J Mol Sci 2019; 20:ijms20102416. [PMID: 31096713 PMCID: PMC6566335 DOI: 10.3390/ijms20102416] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022] Open
Abstract
Classic Hodgkin lymphoma (cHL) is characterized by a few tumor cells surrounded by a protective, immunosuppressive tumor microenvironment composed of normal cells that are an active part of the disease. Hodgkin and Reed-Sternberg (HRS) cells evade the immune system through a variety of different mechanisms. They evade antitumor effector T cells and natural killer cells and promote T cell exhaustion. Using cytokines and extracellular vesicles, they recruit normal cells, induce their proliferation and "educate" (i.e. reprogram) them to become immunosuppressive and protumorigenic. Therefore, alternative treatment strategies are being developed to target not only tumor cells but also the tumor microenvironment. Here we summarize current knowledge on the ability of HRS cells to build their microenvironment and to educate normal cells to become immunosuppressive. We also describe therapeutic strategies to counteract formation of the tumor microenvironment and related processes leading to T cell exhaustion and repolarization of immunosuppressive tumor-associated macrophages.
Collapse
Affiliation(s)
- Donatella Aldinucci
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| | - Cinzia Borghese
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| | - Naike Casagrande
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| |
Collapse
|
11
|
Carreau NA, Diefenbach CS. Immune targeting of the microenvironment in classical Hodgkin's lymphoma: insights for the hematologist. Ther Adv Hematol 2019; 10:2040620719846451. [PMID: 31105921 PMCID: PMC6501496 DOI: 10.1177/2040620719846451] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/02/2019] [Indexed: 12/31/2022] Open
Abstract
While up to 80% of patients with Hodgkin's lymphoma (HL) are cured with first-line therapy, relapsed/refractory (R/R) disease remains a clinical challenge and is fatal for many young patients. HL is unique in that the tumor cells (Hodgkin Reed-Sternberg; HRS cells) are a small fraction (<1%) of the tumor bulk, with the remaining tumor composed of the cells of the tumor microenvironment (TME). The support and integrity of the TME is necessary for HRS cell growth and survival. Targeting the programmed death 1 pathway has shown exciting activity in relapsed HL and led to United States Food and Drug Administration approval of the checkpoint inhibitors, nivolumab and pembrolizumab, for R/R HL. Novel combinations with checkpoint blockade therapy (CBT), targeted approaches such as combinations of CBT with brentuximab vedotin or chemotherapy, chimeric antigen receptor T-cells, and the use of CBT to potentially sensitize to subsequent therapy are being investigated as treatment approaches. As understanding of the HL TME grows, hopefully this will increase the number of rational therapeutic targets.
Collapse
Affiliation(s)
- Nicole A Carreau
- Division of Hematology and Medical Oncology, Perlmutter Cancer Center at NYU Langone Health, New York University School of Medicine & NYU Langone Medical Center, New York, NY, USA
| | - Catherine S Diefenbach
- Division of Hematology and Medical Oncology, Perlmutter Cancer Center at NYU Langone Health, New York University School of Medicine & NYU Langone Medical Center, 240 East 38 Street, 19 Floor, New York, NY 10016, USA
| |
Collapse
|
12
|
Ansell SM. Immunotherapy in Hodgkin Lymphoma: The Road Ahead. Trends Immunol 2019; 40:380-386. [PMID: 30948348 DOI: 10.1016/j.it.2019.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 12/24/2022]
Abstract
An extensive infiltrate of intratumoral immune cells is a hallmark of classic Hodgkin lymphoma (cHL) but these cells do result in an effective antitumor response. Immune checkpoint therapy, which activates 'exhausted' T cells, has been found to be highly effective in cHL, but responding patients commonly relapse. Combination approaches are currently being investigated but the assessment of benefit when adding immunotherapy is challenging. The pitfalls in designing combination studies derive from response endpoints that are difficult to measure, a lack of biomarkers that predict response, and a limited understanding of tumor biology. While progress in treating patients with cHL has been exceptional so far, further progress may require a review of clinical trial endpoints and a greater understanding of cHL biology.
Collapse
Affiliation(s)
- Stephen M Ansell
- Division of Hematology, Mayo Clinic, 200 First Street, Rochester, MN 55905, USA.
| |
Collapse
|
13
|
Anti-PD-1 therapy for clinical treatment of lymphoma: a single-arm meta-analysis. Oncotarget 2018; 9:35343-35355. [PMID: 30450162 PMCID: PMC6219677 DOI: 10.18632/oncotarget.26223] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 06/13/2018] [Indexed: 01/05/2023] Open
Abstract
Anti-PD1 antibodies exhibit satisfactory efficacy in treating certain types of lymphoma. We conducted this meta-analysis to explore subtypes benefiting from this treatment and the best anti-PD1 therapeutic modalities.
Collapse
|
14
|
Dada R, Zabani Y. Nivolumab induces impressive responses in relapsed/refractory classic Hodgkin lymphoma: Single institutional experience. J Oncol Pharm Pract 2018; 25:1586-1589. [PMID: 30253728 DOI: 10.1177/1078155218800150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Cancer can escape the immune system through different mechanisms. One such mechanism is the expression of program death ligand-1 which binds to PD-1 receptor on activated T cells, subsequently leading to inhibition of the immune response against cancer cells. Nivolumab is a novel antibody that binds to PD-1 and prevents such immune tolerance. Two recently published controlled clinical trials confirmed the clinical efficacy of single-agent nivolumab in pretreated patients with classical Hodgkin lymphoma. PATIENTS AND METHODS We treated 10 heavily pretreated patients with classical Hodgkin lymphoma with the new novel PD-1 inhibitor nivolumab. We report on the outcome and safety of this agent in these patients. RESULTS After four cycles, the response rate was 80%. Seven of 10 gained complete metabolic remission. No serious adverse events were observed. The available literature is being reviewed. CONCLUSIONS Pretreated classical Hodgkin lymphoma is amenable to novel immunotherapy. Nivolumab induces clinically meaningful responses with excellent tolerability. The drug enriches our treatment options by reviving the response of the immune system against cancer. Further controlled studies are needed to determine the effectiveness on a large patient cohort.
Collapse
Affiliation(s)
- Reyad Dada
- 1 King Faisal Specialist Hospital and Research Centre, Jeddah, Kingdom of Saudi Arabia.,2 College of Medicine, Al-Faisal University, Riyadh, Kingdom of Saudi Arabia
| | - Yazeed Zabani
- 3 College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
15
|
Stock S, Hoffmann JM, Schubert ML, Wang L, Wang S, Gong W, Neuber B, Gern U, Schmitt A, Müller-Tidow C, Dreger P, Schmitt M, Sellner L. Influence of Retronectin-Mediated T-Cell Activation on Expansion and Phenotype of CD19-Specific Chimeric Antigen Receptor T Cells. Hum Gene Ther 2018; 29:1167-1182. [PMID: 30024314 DOI: 10.1089/hum.2017.237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Enhanced in vivo expansion, long-term persistence of chimeric antigen receptor T (CART) cells, and efficient tumor eradication through these cells are linked to the proportion of less-differentiated cells in the CART cell product. Retronectin is well established as an adjuvant for improved retroviral transduction, while its property to enrich less-differentiated T cells is less known. In order to increase these subsets, this study investigated the effects of retronectin-mediated T-cell activation for CD19-specific CART cell production. Peripheral blood mononuclear cells of healthy donors and untreated chronic lymphocytic leukemia (CLL) patients without or with positive selection for CD3+ T cells were transduced with a CD19.CAR.CD28.CD137zeta third-generation retroviral vector. Activation of peripheral blood mononuclear cells was performed by CD3/CD28, CD3/CD28/retronectin, or CD3/retronectin. Interleukin-7 and -15 were supplemented to all cultures. Retronectin was used in all three activation protocols for retroviral transduction. Expansion was assessed by trypan blue staining. Viability, transduction efficiency, immune phenotype, and cytokine production were longitudinally analyzed by flow cytometry. Cytotoxic capacity of generated CART cells was evaluated using a classical chromium-51 release assay. Retronectin-mediated activation resulted in an enrichment of CD8+ cytotoxic CART cells and less-differentiated naïve-like T cells (CD45RA+CCR7+). Retronectin-activated CART cells showed increased cytotoxic activity. However, activation with retronectin decreased viability, expansion, transduction efficiency, and cytokine production, particularly of CLL patient-derived CART cells. Both retronectin-mediated activation protocols promoted a less-differentiated CART cell phenotype without comprising cytotoxic properties of healthy donor-derived CART cells. However, up-front retronectin resulted in reduced viability and expansion in CLL patients. This effect is probably attributed to the retronectin-mediated activation of B cells with prolonged CLL persistence. Consequently, CART cell expansion and generation failed. In summary, activation with retronectin should be performed with caution and may be limited to patients without a higher percentage of tumor cells in the peripheral blood.
Collapse
Affiliation(s)
- Sophia Stock
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Maria-Luisa Schubert
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Lei Wang
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Sanmei Wang
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Wenjie Gong
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Brigitte Neuber
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Ulrike Gern
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Anita Schmitt
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany
| | - Carsten Müller-Tidow
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| | - Peter Dreger
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| | - Michael Schmitt
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| | - Leopold Sellner
- 1 Department of Medicine V, Heidelberg University Hospital , Heidelberg, Germany; and German Cancer Consortium, Heidelberg, Germany .,2 National Center for Tumor Diseases , German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
16
|
Lamble AJ, Lind EF. Targeting the Immune Microenvironment in Acute Myeloid Leukemia: A Focus on T Cell Immunity. Front Oncol 2018; 8:213. [PMID: 29951373 PMCID: PMC6008423 DOI: 10.3389/fonc.2018.00213] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022] Open
Abstract
Immunotherapies, such as chimeric antigen receptor T cells, bispecific antibodies, and immune checkpoint inhibitors, have emerged as promising modalities in multiple hematologic malignancies. Despite the excitement surrounding immunotherapy, it is currently not possible to predict which patients will respond. Within solid tumors, the status of the immune microenvironment provides valuable insight regarding potential responses to immune therapies. Much less is known about the immune microenvironment within hematologic malignancies but the characteristics of this environment are likely to serve a similar predictive role. Acute myeloid leukemia (AML) is the most common hematologic malignancy in adults, and only 25% of patients are alive 5 years following their diagnosis. There is evidence that manipulation of the immune microenvironment by leukemia cells may play a role in promoting therapy resistance and disease relapse. In addition, it has long been documented that through modulation of the immune system following allogeneic bone marrow transplant, AML can be cured, even in patients with the highest risk disease. These concepts, along with the poor prognosis associated with this disease, have encouraged many groups to start exploring the utility of novel immune therapies in AML. While the implementation of these therapies into clinical trials for AML has been supported by preclinical rationale, many questions still exist surrounding their efficacy, tolerability, and the overall optimal approach. In this review, we discuss what is known about the immune microenvironment within AML with a specific focus on T cells and checkpoints, along with their implications for immune therapies.
Collapse
Affiliation(s)
- Adam J Lamble
- Pediatric Hematology/Oncology, Seattle Children's Hospital, Seattle, WA, United States
| | - Evan F Lind
- Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
17
|
Gravelle P, Burroni B, Péricart S, Rossi C, Bezombes C, Tosolini M, Damotte D, Brousset P, Fournié JJ, Laurent C. Mechanisms of PD-1/PD-L1 expression and prognostic relevance in non-Hodgkin lymphoma: a summary of immunohistochemical studies. Oncotarget 2018; 8:44960-44975. [PMID: 28402953 PMCID: PMC5546533 DOI: 10.18632/oncotarget.16680] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/16/2017] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint blockade therapeutics, notably antibodies targeting the programmed death 1 (PD-1) receptor and its PD-L1 and PD-L2 ligands, are currently revolutionizing the treatment of cancer. For a sizeable fraction of patients with melanoma, lung, kidney and several other solid cancers, monoclonal antibodies that neutralize the interactions of the PD-1/PD-L1 complex allow the reconstitution of long-lasting antitumor immunity. In hematological malignancies this novel therapeutic strategy is far less documented, although promising clinical responses have been seen in refractory and relapsed Hodgkin lymphoma patients. This review describes our current knowledge of PD-1 and PD-L1 expression, as reported by immunohistochemical staining in both non-Hodgkin lymphoma cells and their surrounding immune cells. Here, we discuss the multiple intrinsic and extrinsic mechanisms by which both T and B cell lymphomas up-regulate the PD-1/PD-L1 axis, and review current knowledge about the prognostic significance of its immunohistochemical detection. This body of literature establishes the cell surface expression of PD-1/PD-L1 as a critical determinant for the identification of non-Hodgkin lymphoma patients eligible for immune checkpoint blockade therapies.
Collapse
Affiliation(s)
- Pauline Gravelle
- Département de Pathologie, CHU Toulouse, Institut Universitaire du Cancer de Toulouse, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France.,Institut Universitaire du Cancer de Toulouse, Toulouse, France.,Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| | - Barbara Burroni
- Service de Pathologie Hôpitaux Universitaires Paris Centre, Hopital Cochin, Paris, France
| | - Sarah Péricart
- Département de Pathologie, CHU Toulouse, Institut Universitaire du Cancer de Toulouse, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France.,Institut Universitaire du Cancer de Toulouse, Toulouse, France.,Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| | - Cédric Rossi
- Institut Universitaire du Cancer de Toulouse, Toulouse, France.,Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,CHU le Bocage, Hématologie Clinique, Dijon, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| | - Christine Bezombes
- Institut Universitaire du Cancer de Toulouse, Toulouse, France.,Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| | - Marie Tosolini
- Institut Universitaire du Cancer de Toulouse, Toulouse, France.,Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| | - Diane Damotte
- Service de Pathologie Hôpitaux Universitaires Paris Centre, Hopital Cochin, Paris, France.,Centre de Recherche des Cordeliers, INSERM U1138, Paris, France
| | - Pierre Brousset
- Département de Pathologie, CHU Toulouse, Institut Universitaire du Cancer de Toulouse, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France.,Institut Universitaire du Cancer de Toulouse, Toulouse, France.,Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| | - Camille Laurent
- Département de Pathologie, CHU Toulouse, Institut Universitaire du Cancer de Toulouse, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France.,Institut Universitaire du Cancer de Toulouse, Toulouse, France.,Centre de Recherches en Cancérologie de Toulouse, UMR1037 INSERM-Université Toulouse III, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Institut Carnot CALYM, Toulouse, France.,Paul-Sabatier, ERL 5294 CNRS, Université de Toulouse, Toulouse, France
| |
Collapse
|
18
|
Poggi A, Varesano S, Zocchi MR. How to Hit Mesenchymal Stromal Cells and Make the Tumor Microenvironment Immunostimulant Rather Than Immunosuppressive. Front Immunol 2018; 9:262. [PMID: 29515580 PMCID: PMC5825917 DOI: 10.3389/fimmu.2018.00262] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
Experimental evidence indicates that mesenchymal stromal cells (MSCs) may regulate tumor microenvironment (TME). It is conceivable that the interaction with MSC can influence neoplastic cell functional behavior, remodeling TME and generating a tumor cell niche that supports tissue neovascularization, tumor invasion and metastasization. In addition, MSC can release transforming growth factor-beta that is involved in the epithelial-mesenchymal transition of carcinoma cells; this transition is essential to give rise to aggressive tumor cells and favor cancer progression. Also, MSC can both affect the anti-tumor immune response and limit drug availability surrounding tumor cells, thus creating a sort of barrier. This mechanism, in principle, should limit tumor expansion but, on the contrary, often leads to the impairment of the immune system-mediated recognition of tumor cells. Furthermore, the cross-talk between MSC and anti-tumor lymphocytes of the innate and adaptive arms of the immune system strongly drives TME to become immunosuppressive. Indeed, MSC can trigger the generation of several types of regulatory cells which block immune response and eventually impair the elimination of tumor cells. Based on these considerations, it should be possible to favor the anti-tumor immune response acting on TME. First, we will review the molecular mechanisms involved in MSC-mediated regulation of immune response. Second, we will focus on the experimental data supporting that it is possible to convert TME from immunosuppressive to immunostimulant, specifically targeting MSC.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, Policlinico San Martino, Genoa, Italy
| | - Serena Varesano
- Molecular Oncology and Angiogenesis Unit, Policlinico San Martino, Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
19
|
Nelde A, Walz JS, Kowalewski DJ, Schuster H, Wolz OO, Peper JK, Cardona Gloria Y, Langerak AW, Muggen AF, Claus R, Bonzheim I, Fend F, Salih HR, Kanz L, Rammensee HG, Stevanović S, Weber ANR. HLA class I-restricted MYD88 L265P-derived peptides as specific targets for lymphoma immunotherapy. Oncoimmunology 2016; 6:e1219825. [PMID: 28405493 PMCID: PMC5384368 DOI: 10.1080/2162402x.2016.1219825] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 12/28/2022] Open
Abstract
Genome sequencing has uncovered an array of recurring somatic mutations in different non-Hodgkin lymphoma (NHL) subtypes. If affecting protein-coding regions, such mutations may yield mutation-derived peptides that may be presented by HLA class I proteins and recognized by cytotoxic T cells. A recurring somatic and oncogenic driver mutation of the Toll-like receptor adaptor protein MYD88, Leu265Pro (L265P) was identified in up to 90% of different NHL subtype patients. We therefore screened the potential of MYD88L265P-derived peptides to elicit cytotoxic T cell responses as tumor-specific neoantigens. Based on in silico predictions, we identified potential MYD88L265P-containing HLA ligands for several HLA class I restrictions. A set of HLA class I MYD88L265P-derived ligands elicited specific cytotoxic T cell responses for HLA-B*07 and -B*15. These data highlight the potential of MYD88L265P mutation-specific peptide-based immunotherapy as a novel personalized treatment approach for patients with MYD88L265P+ NHLs that may complement pharmacological approaches targeting oncogenic MyD88 L265P signaling.
Collapse
Affiliation(s)
- Annika Nelde
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Juliane Sarah Walz
- Department of Hematology and Oncology, University Hospital Tübingen, Tübingen, Germany
| | | | - Heiko Schuster
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Olaf-Oliver Wolz
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Janet Kerstin Peper
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Yamel Cardona Gloria
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Anton W. Langerak
- Department of Immunology, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Alice F. Muggen
- Department of Immunology, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Rainer Claus
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, Germany
| | - Irina Bonzheim
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Falko Fend
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Helmut Rainer Salih
- Department of Hematology and Oncology, University Hospital Tübingen, Tübingen, Germany
- Clinical Cooperation Unit Translational Immunology, German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
| | - Lothar Kanz
- Department of Hematology and Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
| | - Alexander N. R. Weber
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
20
|
Poggi A, Giuliani M. Mesenchymal Stromal Cells Can Regulate the Immune Response in the Tumor Microenvironment. Vaccines (Basel) 2016; 4:E41. [PMID: 27834810 PMCID: PMC5192361 DOI: 10.3390/vaccines4040041] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/01/2016] [Accepted: 10/31/2016] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment is a good target for therapy in solid tumors and hematological malignancies. Indeed, solid tumor cells' growth and expansion can influence neighboring cells' behavior, leading to a modulation of mesenchymal stromal cell (MSC) activities and remodeling of extracellular matrix components. This leads to an altered microenvironment, where reparative mechanisms, in the presence of sub-acute inflammation, are not able to reconstitute healthy tissue. Carcinoma cells can undergo epithelial mesenchymal transition (EMT), a key step to generate metastasis; these mesenchymal-like cells display the functional behavior of MSC. Furthermore, MSC can support the survival and growth of leukemic cells within bone marrow participating in the leukemic cell niche. Notably, MSC can inhibit the anti-tumor immune response through either carcinoma-associated fibroblasts or bone marrow stromal cells. Experimental data have indicated their relevance in regulating cytolytic effector lymphocytes of the innate and adaptive arms of the immune system. Herein, we will discuss some of the evidence in hematological malignancies and solid tumors. In particular, we will focus our attention on the means by which it is conceivable to inhibit MSC-mediated immune suppression and trigger anti-tumor innate immunity.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy.
| | - Massimo Giuliani
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City L-1526, Luxembourg.
| |
Collapse
|
21
|
Aldinucci D, Celegato M, Casagrande N. Microenvironmental interactions in classical Hodgkin lymphoma and their role in promoting tumor growth, immune escape and drug resistance. Cancer Lett 2016; 380:243-52. [DOI: 10.1016/j.canlet.2015.10.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 12/22/2022]
|
22
|
Laurent C, Fabiani B, Do C, Tchernonog E, Cartron G, Gravelle P, Amara N, Malot S, Palisoc MM, Copie-Bergman C, Glehen AT, Copin MC, Brousset P, Pittaluga S, Jaffe ES, Coppo P. Immune-checkpoint expression in Epstein-Barr virus positive and negative plasmablastic lymphoma: a clinical and pathological study in 82 patients. Haematologica 2016; 101:976-84. [PMID: 27175027 PMCID: PMC4967577 DOI: 10.3324/haematol.2016.141978] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/03/2016] [Indexed: 12/14/2022] Open
Abstract
Plasmablastic lymphoma is a rare and aggressive diffuse large B-cell lymphoma commonly associated with Epstein-Barr virus co-infection that most often occurs in the context of human immunodeficiency virus infection. Therefore, its immune escape strategy may involve the upregulation of immune-checkpoint proteins allowing the tumor immune evasion. However, the expression of these molecules was poorly studied in this lymphoma. We have investigated 82 plasmablastic lymphoma cases of whom half were Epstein-Barr virus positive. Although they harbored similar pathological features, Epstein-Barr virus positive plasmablastic lymphomas showed a significant increase in MYC gene rearrangement and had a better 2-year event-free survival than Epstein-Barr virus negative cases (P=0.049). Immunostains for programmed cell death-1, programmed cell death-ligand 1, indole 2,3-dioxygenase and dendritic cell specific C-type lectin showed a high or moderate expression by the microenvironment cells in 60%-72% of cases, whereas CD163 was expressed in almost all cases. Tumor cells also expressed programmed cell death-1 and its ligand in 22.5% and 5% of cases, respectively. Both Epstein-Barr virus positive and negative plasmablastic lymphomas exhibited a high immune-checkpoint score showing that it involves several pathways of immune escape. However, Epstein-Barr virus positive lymphomas exhibited a higher expression of programmed cell death-1 and its ligand in both malignant cells and microenvironment as compared to Epstein-Barr virus negative cases. In conclusion, plasmablastic lymphoma expresses immune-checkpoint proteins through both malignant cells and the tumor microenvironment. The expression of programmed cell death-1 and its ligand constitutes a strong rationale for testing monoclonal antibodies in this often chemoresistant disease.
Collapse
Affiliation(s)
- Camille Laurent
- Département de Pathologie, Institut Universitaire du Cancer-Oncopole, Toulouse, France INSERM, U.1037, Centre de Recherche en Cancérologie de Toulouse-Purpan, Toulouse, France
| | - Bettina Fabiani
- Département de Pathologie, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Catherine Do
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | | | - Guillaume Cartron
- Service d'Hematologie, Hôpital Gui de Chauliac-Saint Eloi, Montpellier, France
| | - Pauline Gravelle
- Département de Pathologie, Institut Universitaire du Cancer-Oncopole, Toulouse, France INSERM, U.1037, Centre de Recherche en Cancérologie de Toulouse-Purpan, Toulouse, France
| | - Nadia Amara
- Département de Pathologie, Institut Universitaire du Cancer-Oncopole, Toulouse, France
| | - Sandrine Malot
- Service d'Hématologie, AP-HP, Hôpital Saint-Antoine, Paris, France Centre de Référence des Microangiopathies Thrombotiques, AP-HP, Paris, France
| | | | - Christiane Copie-Bergman
- Département de Pathologie, AP-HP, Groupe Hospitalier Henri Mondor - Albert henevier, Créteil, France
| | | | | | - Pierre Brousset
- Département de Pathologie, Institut Universitaire du Cancer-Oncopole, Toulouse, France INSERM, U.1037, Centre de Recherche en Cancérologie de Toulouse-Purpan, Toulouse, France
| | - Stefania Pittaluga
- Hematopathology Section, Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Elaine S Jaffe
- Hematopathology Section, Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Paul Coppo
- Service d'Hématologie, AP-HP, Hôpital Saint-Antoine, Paris, France Centre de Référence des Microangiopathies Thrombotiques, AP-HP, Paris, France UPMC, Université Paris VI, France Inserm U1170, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
23
|
Anderlini P, Saliba RM, Ledesma C, Plair T, Alousi AM, Hosing CM, Khouri IF, Nieto Y, Popat UR, Shpall EJ, Fanale MA, Hagemeister FB, Oki Y, Neelapu S, Romaguera JE, Younes A, Champlin RE. Gemcitabine, Fludarabine, and Melphalan for Reduced-Intensity Conditioning and Allogeneic Stem Cell Transplantation for Relapsed and Refractory Hodgkin Lymphoma. Biol Blood Marrow Transplant 2016; 22:1333-1337. [PMID: 27064056 PMCID: PMC5193224 DOI: 10.1016/j.bbmt.2016.03.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/27/2016] [Indexed: 01/22/2023]
Abstract
Forty patients (median age, 31 years; range, 20 to 63) with Hodgkin lymphoma underwent an allogeneic stem cell transplant with the gemcitabine-fludarabine-melphalan reduced-intensity conditioning regimen. Thirty-one patients (77%) had undergone a prior autologous stem cell transplant, with a median time to progression after transplant of 6 months (range, 1 to 68). Disease status at transplant was complete remission/complete remission, undetermined (n = 23; 57%), partial remission (n = 14; 35%), and other (n = 3; 8%). Twenty-six patients (65%) received brentuximab vedotin before allotransplant. The overall complete response rate before allotransplant was 65% in brentuximab-treated patients versus 42% in brentuximab-naive patients (P = .15). At the latest follow-up (October 2015) 31 patients were alive. The median follow-up was 41 months (range, 5 to 87). Transplant-related mortality rate at 3 years was 17%. Pulmonary, skin toxicities, and nausea were seen in 13 (33%), 11 (28%), and 37 (93%) patients, respectively. At 3 years, estimates for overall and progression-free survival were 75% (95% CI, 57% to 86%) and 54% (95% CI, 36% to 70%). Overall incidence for disease progression was 28% (95% CI, 16% to 50%). We believe the gemcitabine-fludarabine-melphalan regimen allows moderate dose intensification with acceptable morbidity and mortality. The inclusion of gemcitabine affected nausea, pulmonary, and likely skin toxicity. Exposure to brentuximab vedotin allowed more patients to reach allogeneic stem cell transplantation in complete remission. With over 50% of patients progression-free at 3 years, allogeneic stem cell transplantation with reduced-intensity conditioning remains an effective and relevant treatment option for Hodgkin lymphoma in the brentuximab vedotin era.
Collapse
Affiliation(s)
- Paolo Anderlini
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| | - Rima M Saliba
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Celina Ledesma
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Tamera Plair
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Amin M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Chitra M Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Issa F Khouri
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Uday R Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Michelle A Fanale
- Department of Lymphoma, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Yasuhiro Oki
- Department of Lymphoma, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Saatva Neelapu
- Department of Lymphoma, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Jorge E Romaguera
- Department of Lymphoma, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Anas Younes
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
24
|
Choi MY, Kashyap MK, Kumar D. The chronic lymphocytic leukemia microenvironment: Beyond the B-cell receptor. Best Pract Res Clin Haematol 2016; 29:40-53. [PMID: 27742071 DOI: 10.1016/j.beha.2016.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 07/03/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
Malignant B cells accumulate in the peripheral blood, bone marrow, and lymphoid organs of patients with chronic lymphocytic leukemia (CLL). In the tissue compartments, CLL shape a protective microenvironment by coopting normal elements. The efficacy of drugs that target these interactions further underscores their importance in the pathogenesis of CLL. While the B cell receptor (BCR) pathway clearly plays a central role in the CLL microenvironment, there is also rationale to evaluate agents that inhibit other aspects or modulate the immune cells in the microenvironment. Here we review the main cellular components, soluble factors, and signaling pathways of the CLL microenvironment, and highlight recent clinical advances. As the BCR pathway is reviewed elsewhere, we focus on other aspects of the microenvironment.
Collapse
Affiliation(s)
- Michael Y Choi
- Moores Cancer Center, UCSD-Moores Cancer Center, La Jolla, 92093-0820, CA, USA.
| | - Manoj Kumar Kashyap
- Moores Cancer Center, UCSD-Moores Cancer Center, La Jolla, 92093-0820, CA, USA.
| | - Deepak Kumar
- Moores Cancer Center, UCSD-Moores Cancer Center, La Jolla, 92093-0820, CA, USA.
| |
Collapse
|
25
|
Perales MA, Sauter CS, Armand P. Reprint of: Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2016; 22:S9-S14. [PMID: 26899275 DOI: 10.1016/j.bbmt.2016.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in non-transplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Craig S Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
26
|
Bennani-Baiti N, Thanarajasingam G, Ansell S. Checkpoint Inhibitors for the Treatment of Hodgkin Lymphoma. Expert Rev Clin Immunol 2016; 12:673-9. [PMID: 26818843 DOI: 10.1586/1744666x.2016.1147350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hodgkin lymphoma's (HL) tumor composition is characterized by a paucity of malignant cells and a preponderance of immune and stromal cells. Despite the rich immune milieu within the tumor microenvironment, malignant cells are able to effectively evade the immune system and use immune support to promote lymphoma cell growth and proliferation. Recognizing this has led to the identification of checkpoint inhibitory signals that enable immune evasion and to opening the door to therapeutic strategies on how to exploit the immune system in targeting tumor cells. We discuss herein some of the tumor evasion mechanisms in HL with a particular focus on the immune checkpoint pathways and focus on recent clinical data of checkpoint blockade in HL treatment.
Collapse
Affiliation(s)
| | | | - Stephen Ansell
- a Division of Hematology , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
27
|
Ansell SM. Hodgkin lymphoma: MOPP chemotherapy to PD-1 blockade and beyond. Am J Hematol 2016; 91:109-12. [PMID: 26505486 DOI: 10.1002/ajh.24226] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/20/2015] [Accepted: 10/22/2015] [Indexed: 01/13/2023]
Abstract
Hodgkin lymphoma is a rare lymphoid malignancy affecting ∼9,200 new patients in the United States annually. Progress in the management of this disease over the past 50 years has been remarkable and the prognosis of this malignancy has changed from a uniformly fatal process to one in which the vast majority of patients are expected to be cured. This remarkable progress has been due to the use of combination approaches incorporating chemotherapy and radiation therapy, and now more recently antibody-drug conjugates and immune checkpoint inhibitors. The goal for the future is to develop treatment combinations that successfully treat all patients and markedly decrease the long-term side effects.
Collapse
|
28
|
Abstract
Cancer can escape the immune system through different mechanisms. One of which is the expression of program death ligand-1 (PD-L1). This ligand binds to programmed cell death 1 receptor on activated T cells, subsequently leading to inhibition of the immune response. Nivolumab is a novel antibody that binds to programmed cell death 1 and prevents such immune tolerance. Several recently published clinical trials confirmed the clinical efficacy of single agent nivolumab in pretreated patients with different cancer types. Publications on nivolumab in Hodgkin lymphoma are very scarce. We report on a 30-year-old man with stage IVB Hodgkin lymphoma, who failed nine lines of treatments including high-dose chemotherapy and autologous stem cell transplantation and brentuximab vedotin. He reached a major response after four cycles of nivolumab and got married. The available literature is being reviewed. Pre-treated Hodgkin lymphoma is amenable to novel immunotherapy. Nivolumab induces clinically meaningful responses with excellent tolerance. The drug enriches our treatment options by reviving the immune system response against cancer. Further clinical studies are needed to determine the effectiveness on a large patients' cohort.
Collapse
Affiliation(s)
- Reyad Dada
- 1 King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia.,2 College of Medicine, Al-Faisal University, Riyadh, Kingdom of Saudi Arabia
| | - Jamal Zekri
- 1 King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia.,2 College of Medicine, Al-Faisal University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
29
|
Martino M, Festuccia M, Fedele R, Console G, Cimminiello M, Gavarotti P, Bruno B. Salvage treatment for relapsed/refractory Hodgkin lymphoma: role of allografting, brentuximab vedotin and newer agents. Expert Opin Biol Ther 2015; 16:347-64. [PMID: 26652934 DOI: 10.1517/14712598.2015.1130821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Second-line, salvage chemotherapy followed by high-dose chemotherapy and autologous stem cell transplantation (AUTO-SCT) is the standard of care for patients with relapsed/refractory (R/R) Hodgkin lymphoma (HL). Approximately 50% of patients relapse after AUTO-SCT and their prognosis is generally poor. Brentuximab Vedotin (BV) has demonstrated efficacy in this setting and allogeneic (ALLO)-SCT represents an option with curative potential in this subgroup of patients. AREAS COVERED A systematic review has been conducted to explore the actual knowledge on ALLO-SCT, BV and newer agents in R/R HL. EXPERT OPINION The introduction of BV in clinical practice has significantly improved the management of post-AUTO-SCT relapses and the drug can induce durable remissions in a subset of R/R HL. Allografting select patients has been used to improve clinical outcomes and recent case series have begun to explore BV as a potential 'bridge' to allo-SCT, even though the optimal timing of ALLO-SCT after BV response remains undetermined. However, reduced tumor burden at the time of ALLO-SCT is a key factor to decrease relapse risk. Based on the unique composition of the tumor, more recently new agents such as PD-1 inhibitors have been developed. The potential role of PD-1 inhibitors with ALLO-SCT remains to be explored.
Collapse
Affiliation(s)
- Massimo Martino
- a Hematology and Stem Cells Transplantation Unit , CTMO, Azienda Ospedaliera 'BMM' , Reggio , Italy
| | - Moreno Festuccia
- b Division of Hematology, A.O.U. Citta' della Salute e della Scienza di Torino - Presidio Molinette, and Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| | - Roberta Fedele
- a Hematology and Stem Cells Transplantation Unit , CTMO, Azienda Ospedaliera 'BMM' , Reggio , Italy
| | - Giuseppe Console
- a Hematology and Stem Cells Transplantation Unit , CTMO, Azienda Ospedaliera 'BMM' , Reggio , Italy
| | - Michele Cimminiello
- c Hematology and Stem Cell Transplant Unit , Azienda Ospedaliera San Carlo , Potenza , Italy
| | - Paolo Gavarotti
- b Division of Hematology, A.O.U. Citta' della Salute e della Scienza di Torino - Presidio Molinette, and Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| | - Benedetto Bruno
- b Division of Hematology, A.O.U. Citta' della Salute e della Scienza di Torino - Presidio Molinette, and Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| |
Collapse
|
30
|
Rueda Domínguez A, Alfaro Lizaso J, de la Cruz Merino L, Gumá I Padró J, Quero Blanco C, Gómez Codina J, Llanos Muñoz M, Martinez Banaclocha N, Rodriguez Abreu D, Provencio Pulla M. SEOM clinical guidelines for the treatment of Hodgkin's lymphoma. Clin Transl Oncol 2015; 17:1005-13. [PMID: 26497354 PMCID: PMC4689754 DOI: 10.1007/s12094-015-1429-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 10/09/2015] [Indexed: 11/27/2022]
Abstract
Hodgkin lymphoma (HL) is an uncommon B cell lymphoid malignancy representing approximately 10-15 % of all lymphomas. HL is composed of two distinct disease entities; the more commonly diagnosed classical HL and the rare nodular lymphocyte-predominant HL. An accurate assessment of the stage of disease and prognostic factors that identify patients at low or high risk for recurrence are used to optimize therapy. Patients with early stage disease are treated with combined modality strategies using abbreviated courses of combination chemotherapy followed by involved-field radiation therapy, while those with advanced stage disease receive a longer course of chemotherapy often without radiation therapy. High-dose chemotherapy (HDCT) followed by an autologous stem cell transplant (ASCT) is the standard of care for most patients who relapse following initial therapy. Brentuximab vedotin should be considered for patients who fail HDCT with ASCT.
Collapse
Affiliation(s)
- A Rueda Domínguez
- Área de Oncología y Hematología, Hospital Costa del Sol, Autovía A-7, km 187, 29603, Marbella, Málaga, Spain.
| | - J Alfaro Lizaso
- Servicio de Oncología Médica, Instituto Oncológico de Guipúzcoa, San Sebastian, Spain
| | - L de la Cruz Merino
- Servicio de Oncología Médica, Complejo Hospitalario Regional Virgen Macarena, Seville, Spain
| | - J Gumá I Padró
- Servicio de Oncología Médica, Hospital Universitari de Sant Joan de Reus, Reus, Spain
| | - C Quero Blanco
- Servicio de Oncología Médica, Complejo Hospitalario Regional y Virgen de la Victoria, Málaga, Spain
| | - J Gómez Codina
- Servicio de Oncología Médica, Hospital Universitari i Politècnic la Fe, Valencia, Spain
| | - M Llanos Muñoz
- Servicio de Oncología Médica, Hospital Universitario de Canarias (H.U.C), San Cristóbal De La Laguna, Tenerife, Spain
| | - N Martinez Banaclocha
- Servicio de Oncología Médica, Hospital General Universitario de Elche y Vega Baja, Elche, Spain
| | - D Rodriguez Abreu
- Servicio de Oncología Médica, Hospital Universitario Insular de Gran Canaria, Las Palmas De Gran Canarias, Spain
| | - M Provencio Pulla
- Servicio de Oncología Médica, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
31
|
Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2015; 22:17-22. [PMID: 26485445 DOI: 10.1016/j.bbmt.2015.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 01/21/2023]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in nontransplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
|
32
|
Derer A, Deloch L, Rubner Y, Fietkau R, Frey B, Gaipl US. Radio-Immunotherapy-Induced Immunogenic Cancer Cells as Basis for Induction of Systemic Anti-Tumor Immune Responses - Pre-Clinical Evidence and Ongoing Clinical Applications. Front Immunol 2015; 6:505. [PMID: 26500646 PMCID: PMC4597129 DOI: 10.3389/fimmu.2015.00505] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/16/2015] [Indexed: 01/18/2023] Open
Abstract
Radiotherapy (RT) primarily aims to locally destroy the tumor via the induction of DNA damage in the tumor cells. However, the so-called abscopal, namely systemic and immune–mediated, effects of RT move over more and more in the focus of scientists and clinicians since combinations of local irradiation with immune therapy have been demonstrated to induce anti-tumor immunity. We here summarize changes of the phenotype and microenvironment of tumor cells after exposure to irradiation, chemotherapeutic agents, and immune modulating agents rendering the tumor more immunogenic. The impact of therapy-modified tumor cells and damage-associated molecular patterns on local and systemic control of the primary tumor, recurrent tumors, and metastases will be outlined. Finally, clinical studies affirming the bench-side findings of interactions and synergies of radiation therapy and immunotherapy will be discussed. Focus is set on combination of radio(chemo)therapy (RCT) with immune checkpoint inhibitors, growth factor inhibitors, and chimeric antigen receptor T-cell therapy. Well-deliberated combination of RCT with selected immune therapies and growth factor inhibitors bear the great potential to further improve anti-cancer therapies.
Collapse
Affiliation(s)
- Anja Derer
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Lisa Deloch
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Yvonne Rubner
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|